1
|
Inchingolo AM, Inchingolo AD, Nardelli P, Latini G, Trilli I, Ferrante L, Malcangi G, Palermo A, Inchingolo F, Dipalma G. Stem Cells: Present Understanding and Prospects for Regenerative Dentistry. J Funct Biomater 2024; 15:308. [PMID: 39452606 PMCID: PMC11508604 DOI: 10.3390/jfb15100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Regenerative medicine in dentistry focuses on repairing damaged oral tissues using advanced tools like stem cells, biomaterials, and tissue engineering (TE). Mesenchymal stem cells (MSCs) from dental sources, such as dental pulp and periodontal ligament, show significant potential for tissue regeneration due to their proliferative and differentiative abilities. This systematic review, following PRISMA guidelines, evaluated fifteen studies and identified effective strategies for improving dental, periodontal, and bone tissue regeneration through scaffolds, secretomes, and bioengineering methods. Key advancements include the use of dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) to boost cell viability and manage inflammation. Additionally, pharmacological agents like matrine and surface modifications on biomaterials improve stem cell adhesion and promote osteogenic differentiation. By integrating these approaches, regenerative medicine and TE can optimize dental therapies and enhance patient outcomes. This review highlights the potential and challenges in this field, providing a critical assessment of current research and future directions.
Collapse
Affiliation(s)
- Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Paola Nardelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giulia Latini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Irma Trilli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| |
Collapse
|
2
|
Sarkar R, Paul D, Chatterjee A, Bhattacharya A, Pradhan S, Goswami RK, Sen P. Unveiling the anticancer potential of Pestalotioprolide E, an unexplored macrolide: Targeting TRXR1-TRX1-ASK1-P38 signaling cascade in triple-negative breast cancer. Toxicol In Vitro 2024; 100:105920. [PMID: 39173682 DOI: 10.1016/j.tiv.2024.105920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 08/02/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and metastatic in nature. Existing treatment modalities for TNBC are associated with severe side effects. Thioredoxin reductase (TRXR), the pivotal component of the thioredoxin system, remains overexpressed in various cancer cells including TNBC; promotes cell growth, proliferation, and metastasis, and inhibits apoptosis. Pestalotioprolide E is one of the potent macrolides, a class of secondary metabolites derived from an endophytic fungus Pestalotiopsis microspora with relatively unexplored biological activities. Our study revealed increased expression and activity of TRXR1 in MDA-MB-231 cells compared to the non-cancerous cells. In silico docking analysis and in vitro activity assay demonstrated that Pestalotioprolide E directly interacts with TRXR1 and inhibits its enzymatic activity. This inhibition induces apoptosis via TRX1/ASK1/P38MAPK death signaling cascade and retards metastasis through modulating VEGF, MMP-2, MMP-9, E-cadherin, N-cadherin in MDA-MB-231 cells. Taken together present study establishes TRXR1 as a molecular target for Pestalotioprolide E and its anticancer effect can be attributed to the inhibition of TRXR1 activity in MDA-MB-231.
Collapse
Affiliation(s)
- Ruma Sarkar
- School of Biological Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India; B. D. Patel Institute of Paramedical Sciences, Charotar University of Science and Technology, CHARUSAT Campus, Changa 388421, Gujarat, India
| | - Debobrata Paul
- School of Chemical Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Akash Chatterjee
- School of Biological Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Anindita Bhattacharya
- School of Biological Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Sayantan Pradhan
- School of Biological Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Rajib Kumar Goswami
- School of Chemical Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India.
| | - Prosenjit Sen
- School of Biological Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
3
|
Park HK, Choi YD, Shim HJ, Choi Y, Chung IJ, Yun SJ. Comparative Whole-Genome Sequencing Analysis of In-situ and Invasive Acral Lentiginous Melanoma: Markedly Increased Copy Number Gains of GAB2 , PAK1 , UCP2 , and CCND1 are Associated with Melanoma Invasion. Am J Surg Pathol 2024; 48:1061-1071. [PMID: 38916228 DOI: 10.1097/pas.0000000000002273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Acral lentiginous melanoma (ALM) is the most common subtype of acral melanoma. Even though recent genetic studies are reported in acral melanomas, the genetic differences between in-situ and invasive ALM remain unclear. We aimed to analyze specific genetic changes in ALM and compare genetic differences between in-situ and invasive lesions to identify genetic changes associated with the pathogenesis and progression of ALM. We performed whole genome sequencing of 71 tissue samples from 29 patients with ALM. Comparative analyses were performed, pairing in-situ ALMs with normal tissues and, furthermore, invasive ALMs with normal and in-situ tissues. Among 21 patients with in-situ ALMs, 3 patients (14.3%) had SMIM14 , SLC9B1 , FRG1 , FAM205A , ESRRA , and ESPN mutations, and copy number (CN) gains were identified in only 2 patients (9.5%). Comparing 13 invasive ALMs with in-situ tissues, CN gains were identified in GAB2 in 8 patients (61.5%), PAK1 in 6 patients (46.2%), and UCP2 and CCND1 in 5 patients (38.5%). Structural variants were frequent in in-situ and invasive ALM lesions. Both in-situ and invasive ALMs had very low frequencies of common driver mutations. Structural variants were common in both in-situ and invasive ALMs. Invasive ALMs had markedly increased CN gains, such as GAB2 , PAK1 , UCP2 , and CCND1 , compared with in-situ lesions. These results suggest that they are associated with melanoma invasion.
Collapse
Affiliation(s)
| | | | - Hyun Jeong Shim
- Internal medicine, Division of Hemato-Oncology, Chonnam National University Medical School, Gwangju
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Korea
| | - Ik Joo Chung
- Internal medicine, Division of Hemato-Oncology, Chonnam National University Medical School, Gwangju
| | | |
Collapse
|
4
|
Javakhishvili I, Mardaleishvili K, Buleishvili M, Mantskava M, Chkhikvishvili I, Kalmakhelidze S, Kipiani N, Sanikidze T. Possible link between familial susceptibility to cancer and the level of oxidative stress in thyroid cancer patients. Hered Cancer Clin Pract 2024; 22:15. [PMID: 39180118 PMCID: PMC11342469 DOI: 10.1186/s13053-024-00287-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Hereditary cancer is estimated to account for up to 10% of the worldwide cancer burden; 5% of all thyroid cancers are thought to be genetic. Inheritance of a deleterious mutation in genes associated with a high lifetime risk of developing cancer. Cancer-predisposing genes can promote the initiation and progression of thyroid cancer by enhancing the activation of major signaling pathways through oxidative stress mechanisms. AIM Identification of the possible link between familial susceptibility to cancer and the level of oxidative stress in thyroid cancer patients. METHODS Patients with thyroid cancer (with and without genetic predisposition) were investigated. Study participants were treated in Limited Liability Company (LLC) "Oncology Scientific Research Center" (Tbilisi, Georgia). The study group was collected between 2020 and 2021. In patients' blood, the thyroid hormones content (free Triiodothyronine (fFT3), free Thyroxine (fFT4), bound Triiodothyronine (FT3), bound Thyroxine (FT4), Thyroid-stimulating hormone (TSH)), and oxidative stress intensity (total activity of non-enzymatic antioxidant system (TAA) and the lipid peroxidation product, malondialdehyde (MDA), content) were investigated. RESULTS The difference in free and bound forms of T3 and T4 levels in the blood serum between patients with thyroid cancer (Group 2 and Group 3) and the control group (Group 1) was not statistically significant (F1,2=0.5, p1,2=0.8, F1,3=2.31, p1,3=0.16). In patients with thyroid cancer the TSH level significantly increased compared to the control group (Group 1) (TSH (mean ± Std error): Group 1- 1.21 ± 0.12, Group 2-2.45 ± 0.11 (F1,2=107, p1,2<0.001), Group 3-2.47 ± 0.17 (F1,3=150, p1,3<0.001)) and the MDA levels increased by 4-5 fold. In patients with thyroid cancer from families with cancer aggregation(Group 2), the level of TAA statistically significantly decreased (F1 - 2=200; p1 - 2<0.001), in patients without genetic predisposition to cancer(Group 3), the level of TAA did not change compared to the control (F1 - 3= 2.13; p1 - 3=0.15), CONCLUSIONS: Oxidative stress plays a critical role in tumorigenesis, and antioxidant/oxidant imbalance may contribute to the malignant transformation of normal tissue. In patients with familial susceptibility to cancer mutations of several genes, which are involved in the regulation of oxidative metabolism, may contribute to the disruption of the redox balance, increase the level of oxidative stress, and contribute to the development of thyroid cancer.
Collapse
Affiliation(s)
| | | | - Maka Buleishvili
- Department of Physics, Biophysics, Biomechanics and Informative Technologies of Tbilisi, State Medical University, Tbilisi, Georgia
- Caucasus International University, Tbilisi, Georgia
| | - Maia Mantskava
- Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | | | - Sophio Kalmakhelidze
- Beritashvili Center of Experimental Biomedicine, Tbilisi State Medical University, Tbilisi, Georgia
| | - Nina Kipiani
- Beritashvili Center of Experimental Biomedicine, Tbilisi State Medical University, Tbilisi, Georgia
| | - Tamar Sanikidze
- Department of Physics, Biophysics, Biomechanics and Informative Technologies of Tbilisi, State Medical University, Tbilisi, Georgia.
| |
Collapse
|
5
|
Cesca BA, Caverzan MD, Lamberti MJ, Ibarra LE. Enhancing Therapeutic Approaches in Glioblastoma with Pro-Oxidant Treatments and Synergistic Combinations: In Vitro Experience of Doxorubicin and Photodynamic Therapy. Int J Mol Sci 2024; 25:7525. [PMID: 39062770 PMCID: PMC11277534 DOI: 10.3390/ijms25147525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive brain cancer characterized by significant molecular and cellular heterogeneity, which complicates treatment efforts. Current standard therapies, including surgical resection, radiation, and temozolomide (TMZ) chemotherapy, often fail to achieve long-term remission due to tumor recurrence and resistance. A pro-oxidant environment is involved in glioma progression, with oxidative stress contributing to the genetic instability that leads to gliomagenesis. Evaluating pro-oxidant therapies in brain tumors is crucial due to their potential to selectively target and eradicate cancer cells by exploiting the elevated oxidative stress levels inherent in these malignant cells, thereby offering a novel and effective strategy for overcoming resistance to conventional therapies. This study investigates the therapeutic potential of doxorubicin (DOX) and photodynamic therapy (PDT) with Me-ALA, focusing on their effects on redox homeostasis. Basal ROS levels and antioxidant gene expression (NFE2L2, CAT, GSR) were quantitatively assessed across GBM cell lines, revealing significant variability probably linked to genetic differences. DOX and PDT treatments, both individually and in combination, were analyzed for their efficacy in inducing oxidative stress and cytotoxicity. An in silico analysis further explored the relationship between gene mutations and oxidative stress in GBM patients, providing insights into the molecular mechanisms underlying treatment responses. Our findings suggest that pro-oxidant therapies, such as DOX and PDT in combination, could selectively target GBM cells, highlighting a promising avenue for improving therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Bruno Agustín Cesca
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
| | - Matías Daniel Caverzan
- Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina;
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - María Julia Lamberti
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - Luis Exequiel Ibarra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| |
Collapse
|
6
|
Ionescu C, Kamal FZ, Ciobica A, Halitchi G, Burlui V, Petroaie AD. Oxidative Stress in the Pathogenesis of Oral Cancer. Biomedicines 2024; 12:1150. [PMID: 38927357 PMCID: PMC11200501 DOI: 10.3390/biomedicines12061150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Oxidative stress, arising from an imbalance between reactive oxygen species (ROS) and antioxidants, contributes significantly to oral cancer such as oral squamous cell carcinoma (OSCC) initiation, promotion, and progression. ROS, generated both internally and externally, induce cellular damage including DNA mutations and lipid peroxidation, fostering oncogene activation and carcinogenesis. The objective of this review was to cover and analyze the interplay between ROS and antioxidants, influencing the key processes such as cell proliferation, apoptosis, and angiogenesis, shaping the trajectory of OSCC development. Despite the promise of antioxidants to halt cancer progression and mitigate oxidative damage, their therapeutic efficacy remains debated. The conducted literature search highlighted potential biomarkers that indicate levels of oxidative stress, showing promise for the early detection and monitoring of OSCC. Furthermore, melatonin has emerged as a promising adjunct therapy for OSCC, exerting antioxidant and oncostatic effects by modulating tumor-associated neutrophils and inhibiting cancer cell survival and migration. In addition, this review aims to shed light on developing personalized therapeutic strategies for patients with OSCC such as melatonin therapy, which will be discussed. Research is needed to elucidate the underlying mechanisms and clinical implications of oxidative stress modulation in the context of oral cancer.
Collapse
Affiliation(s)
- Cătălina Ionescu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, Bd. Carol I no. 20A, 700505 Iasi, Romania; (C.I.); (A.C.)
- Clinical Department, Apollonia University, Păcurari Street 11, 700511 Iasi, Romania;
| | - Fatima Zahra Kamal
- Higher Institute of Nursing Professions and Health Technical (ISPITS), Marrakech 40000, Morocco
- Laboratory of Physical Chemistry of Processes and Materials, Faculty of Sciences and Techniques, Hassan First University, B.P. 539, Settat 26000, Morocco
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, Bd. Carol I no. 20A, 700505 Iasi, Romania; (C.I.); (A.C.)
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Teodor Codrescu 2, 700481 Iasi, Romania
- Academy of Romanian Scientists, Str. Splaiul Independentei no. 54, Sector 5, 050094 Bucharest, Romania
| | - Gabriela Halitchi
- Clinical Department, Apollonia University, Păcurari Street 11, 700511 Iasi, Romania;
| | - Vasile Burlui
- Clinical Department, Apollonia University, Păcurari Street 11, 700511 Iasi, Romania;
- Academy of Romanian Scientists, Str. Splaiul Independentei no. 54, Sector 5, 050094 Bucharest, Romania
| | - Antoneta Dacia Petroaie
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| |
Collapse
|
7
|
Li M, Deng X, Zhou D, Liu X, Dai J, Liu Q. A Novel Methylation-based Model for Prognostic Prediction in Lung Adenocarcinoma. Curr Genomics 2024; 25:26-40. [PMID: 38544827 PMCID: PMC10964088 DOI: 10.2174/0113892029277397231228062412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 08/25/2024] Open
Abstract
Objectives Specific methylation sites have shown promise in the early diagnosis of lung adenocarcinoma (LUAD). However, their utility in predicting LUAD prognosis remains unclear. This study aimed to construct a reliable methylation-based predictor for accurately predicting the prognosis of LUAD patients. Methods DNA methylation data and survival data from LUAD patients were obtained from the TCGA and a GEO series. A DNA methylation-based signature was developed using univariate least absolute shrinkage and selection operators and multivariate Cox regression models. Results Eight CpG sites were identified and validated as optimal prognostic signatures for the overall survival of LUAD patients. Receiver operating characteristic analysis demonstrated the high predictive ability of the eight-site methylation signature combined with clinical factors for overall survival. Conclusion This research successfully identified a novel eight-site methylation signature for predicting the overall survival of LUAD patients through bioinformatic integrated analysis of gene methylation markers used in the early diagnosis of lung cancer.
Collapse
Affiliation(s)
- Manyuan Li
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xufeng Deng
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Dong Zhou
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xiaoqing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jigang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Quanxing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| |
Collapse
|
8
|
Shin E, Kwon Y, Jung E, Kim YJ, Kim C, Hong S, Kim J. TM4SF19 controls GABP-dependent YAP transcription in head and neck cancer under oxidative stress conditions. Proc Natl Acad Sci U S A 2024; 121:e2314346121. [PMID: 38315837 PMCID: PMC10873613 DOI: 10.1073/pnas.2314346121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
Tobacco and alcohol are risk factors for human papillomavirus-negative head and neck squamous cell carcinoma (HPV- HNSCC), which arises from the mucosal epithelium of the upper aerodigestive tract. Notably, despite the mutagenic potential of smoking, HPV- HNSCC exhibits a low mutational load directly attributed to smoking, which implies an undefined role of smoking in HPV- HNSCC. Elevated YAP (Yes-associated protein) mRNA is prevalent in HPV- HNSCC, irrespective of the YAP gene amplification status, and the mechanism behind this upregulation remains elusive. Here, we report that oxidative stress, induced by major risk factors for HPV- HNSCC such as tobacco and alcohol, promotes YAP transcription via TM4SF19 (transmembrane 4 L six family member 19). TM4SF19 modulates YAP transcription by interacting with the GABP (Guanine and adenine-binding protein) transcription factor complex. Mechanistically, oxidative stress induces TM4SF19 dimerization and topology inversion in the endoplasmic reticulum membrane, which in turn protects the GABPβ1 subunit from proteasomal degradation. Conversely, depletion of TM4SF19 impairs the survival, proliferation, and migration of HPV- HNSCC cells, highlighting the potential therapeutic relevance of targeting TM4SF19. Our findings reveal the roles of the key risk factors of HPV- HNSCC in tumor development via oxidative stress, offering implications for upcoming therapeutic approaches in HPV- HNSCC.
Collapse
Affiliation(s)
- Eunbie Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Yongsoo Kwon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Eunji Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Yong Joon Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul03722, South Korea
| | - Changgon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Semyeong Hong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| |
Collapse
|
9
|
Hong X, Fu R. Construction of a 5-gene prognostic signature based on oxidative stress related genes for predicting prognosis in osteosarcoma. PLoS One 2023; 18:e0295364. [PMID: 38039294 PMCID: PMC10691720 DOI: 10.1371/journal.pone.0295364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND The understanding of the complex biological scenario of osteosarcoma will open the way to identifying new strategies for its treatment. Oxidative stress is a cancer-related biological scenario. At present, it is not clear the oxidative stress genes in affecting the prognosis and progression of osteosarcoma, the underlying mechanism as well as their impact on the classification of osteosarcoma subtypes. METHODS We selected samples and sequencing data from TARGET data set and GSE21257 data set, and downloaded oxidative stress related-genes (OSRGs) from MsigDB. Univariate Cox analysis of OSRG was conducted using TARGET data, and the prognostic OSRG was screened to conduct unsupervised clustering analysis to identify the molecular subtypes of osteosarcoma. Through least absolute shrinkage and selection operator (LASSO) regression analysis and COX regression analysis of differentially expressed genes (DEGs) between subgroups, a risk assessment system for osteosarcoma was developed. RESULTS 45 prognosis-related OSRGs genes were acquired, and two molecular subtypes of osteosarcoma were clustered. C2 cluster displayed prolonged overall survival (OS) accompanied with high degree of immune infiltration and enriched immune pathways. While cell cycle related pathways were enriched in C2 cluster. Based on DEGs between subgroups and Lasso analysis, 5 hub genes (ZYX, GJA5, GAL, GRAMD1B, and CKMT2) were screened to establish a robust prognostic risk model independent of clinicopathological features. High-risk group had more patients with cancer metastasis and death as well as C1 subtype with poor prognosis. Low-risk group exhibited favorable OS and high immune infiltration status. Additionally, the risk assessment system was optimized by building decision tree and nomogram. CONCLUSIONS This study defined two molecular subtypes of osteosarcoma with different prognosis and tumor immune microenvironment status based on the expression of OSRGs, and provided a new risk assessment system for the prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Xiaofang Hong
- Department of Stomatology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Ribin Fu
- Department of Joint Surgery and Sports Medicine, Zhongshan Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
10
|
Wei T, Liu J, Ma S, Wang M, Yuan Q, Huang A, Wu Z, Shang D, Yin P. A Nucleotide Metabolism-Related Gene Signature for Risk Stratification and Prognosis Prediction in Hepatocellular Carcinoma Based on an Integrated Transcriptomics and Metabolomics Approach. Metabolites 2023; 13:1116. [PMID: 37999212 PMCID: PMC10673507 DOI: 10.3390/metabo13111116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 11/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. The in-depth study of genes and metabolites related to nucleotide metabolism will provide new ideas for predicting the prognosis of HCC patients. This study integrated the transcriptome data of different cancer types to explore the characteristics and significance of nucleotide metabolism-related genes (NMGRs) in different cancer types. Then, we constructed a new HCC classifier and prognosis model based on HCC samples from TCGA and GEO, and detected the gene expression level in the model through molecular biology experiments. Finally, nucleotide metabolism-related products in serum of HCC patients were examined using untargeted metabolomics. A total of 97 NMRGs were obtained based on bioinformatics techniques. In addition, a clinical model that could accurately predict the prognostic outcome of HCC was constructed, which contained 11 NMRGs. The results of PCR experiments showed that the expression levels of these genes were basically consistent with the predicted trends. Meanwhile, the results of untargeted metabolomics also proved that there was a significant nucleotide metabolism disorder in the development of HCC. Our results provide a promising insight into nucleotide metabolism in HCC, as well as a tailored prognostic and chemotherapy sensitivity prediction tool for patients.
Collapse
Affiliation(s)
- Tianfu Wei
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (T.W.)
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Jifeng Liu
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (T.W.)
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Shurong Ma
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (T.W.)
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Mimi Wang
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Qihang Yuan
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (T.W.)
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Anliang Huang
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Zeming Wu
- iPhenome Biotechnology (Yun Pu Kang) Inc., Dalian 116000, China
| | - Dong Shang
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (T.W.)
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Peiyuan Yin
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (T.W.)
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
11
|
Liu M, Wang W, Zhang H, Bi J, Zhang B, Shi T, Su G, Zheng Y, Fan S, Huang X, Chen B, Song Y, Zhao Z, Shi J, Li P, Lu W, Zhang L. Three-Dimensional Gene Regulation Network in Glioblastoma Ferroptosis. Int J Mol Sci 2023; 24:14945. [PMID: 37834393 PMCID: PMC10574000 DOI: 10.3390/ijms241914945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/30/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Ferroptosis is an iron-dependent form of cell death, which is reported to be associated with glioma progression and drug sensitivity. Targeting ferroptosis is a potential therapeutic approach for glioma. However, the molecular mechanism of glioma cell ferroptosis is not clear. In this study, we profile the change of 3D chromatin structure in glioblastoma ferroptosis by using HiChIP and study the 3D gene regulation network in glioblastoma ferroptosis. A combination of an analysis of HiChIP and RNA-seq data suggests that change of chromatin loops mediated by 3D chromatin structure regulates gene expressions in glioblastoma ferroptosis. Genes that are regulated by 3D chromatin structures include genes that were reported to function in ferroptosis, like HDM2 and TXNRD1. We propose a new regulatory mechanism governing glioblastoma cell ferroptosis by 3D chromatin structure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Wange Lu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.L.); (W.W.); (H.Z.); (J.B.); (B.Z.); (T.S.); (G.S.); (Y.Z.); (S.F.); (X.H.); (B.C.); (Y.S.); (Z.Z.); (J.S.); (P.L.)
| | - Lei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.L.); (W.W.); (H.Z.); (J.B.); (B.Z.); (T.S.); (G.S.); (Y.Z.); (S.F.); (X.H.); (B.C.); (Y.S.); (Z.Z.); (J.S.); (P.L.)
| |
Collapse
|
12
|
Nisco A, Carvalho TMA, Tolomeo M, Di Molfetta D, Leone P, Galluccio M, Medina M, Indiveri C, Reshkin SJ, Cardone RA, Barile M. Increased demand for FAD synthesis in differentiated and stem pancreatic cancer cells is accomplished by modulating FLAD1 gene expression: the inhibitory effect of Chicago Sky Blue. FEBS J 2023; 290:4679-4694. [PMID: 37254652 DOI: 10.1111/febs.16881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/31/2023] [Accepted: 05/30/2023] [Indexed: 06/01/2023]
Abstract
FLAD1, along with its FAD synthase (FADS, EC 2.7.7.2) product, is crucial for flavin homeostasis and, due to its role in the mitochondrial respiratory chain and nuclear epigenetics, is closely related to cellular metabolism. Therefore, it is not surprising that it could be correlated with cancer. To our knowledge, no previous study has investigated FLAD1 prognostic significance in pancreatic ductal adenocarcinoma (PDAC). Thus, in the present work, the FAD synthesis process was evaluated in two PDAC cell lines: (a) PANC-1- and PANC-1-derived cancer stem cells (CSCs), presenting the R273H mutation in the oncosuppressor p53, and (b) MiaPaca2 and MiaPaca2-derived CSCs, presenting the R248W mutation in p53. As a control, HPDE cells expressing wt-p53 were used. FADS expression/activity increase was found with malignancy and even more with stemness. An increased FAD synthesis rate in cancer cell lines is presumably demanded by the increase in the FAD-dependent lysine demethylase 1 protein amount as well as by the increased expression levels of the flavoprotein subunit of complex II of the mitochondrial respiratory chain, namely succinate dehydrogenase. With the aim of proposing FADS as a novel target for cancer therapy, the inhibitory effect of Chicago Sky Blue on FADS enzymatic activity was tested on the recombinant 6His-hFADS2 (IC50 = 1.2 μm) and PANC-1-derived CSCs' lysate (IC50 = 2-10 μm). This molecule was found effective in inhibiting the growth of PANC-1 and even more of its derived CSC line, thus assessing its role as a potential chemotherapeutic drug.
Collapse
Affiliation(s)
- Alessia Nisco
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Tiago M A Carvalho
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Piero Leone
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Milagros Medina
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) (GBsC-CSIC Joint Unit), University of Zaragoza, Spain
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Barile
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| |
Collapse
|
13
|
Chen X, Tang Y, Wu D, Li R, Lin Z, Zhou X, Wang H, Zhai H, Xu J, Shi X, Zhang G. From imaging to clinical outcome: dual-region CT radiomics predicting FOXM1 expression and prognosis in hepatocellular carcinoma. Front Oncol 2023; 13:1278467. [PMID: 37817774 PMCID: PMC10561750 DOI: 10.3389/fonc.2023.1278467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Background Liver cancer, especially hepatocellular carcinoma (HCC), remains a significant global health challenge. Traditional prognostic indicators for HCC often fall short in providing comprehensive insights for individualized treatment. The integration of genomics and radiomics offers a promising avenue for enhancing the precision of HCC diagnosis and prognosis. Methods From the Cancer Genome Atlas (TCGA) database, we categorized mRNA of HCC patients by Forkhead Box M1 (FOXM1) expression and performed univariate and multivariate studies to pinpoint autonomous HCC risk factors. We deployed subgroup, correlation, and interaction analyses to probe FOXM1's link with clinicopathological elements. The connection between FOXM1 and immune cells was evaluated using the CIBERSORTx database. The functions of FOXM1 were investigated through analyses of Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). After filtering through TCGA and the Cancer Imaging Archive (TCIA) database, we employed dual-region computed tomography (CT) radiomics technology to noninvasively predict the mRNA expression of FOXM1 in HCC tissues. Radiomic features were extracted from both tumoral and peritumoral regions, and a radiomics score (RS) was derived. The performance and robustness of the constructed models were evaluated using 10-fold cross-validation. A radiomics nomogram was developed by incorporating RS and clinical variables from the TCGA database. The models' discriminative abilities were assessed using metrics such as the area under the curve (AUC) of the receiver operating characteristic curves (ROC) and precision-recall (PR) curves. Results Our findings emphasized the overexpression of FOXM1 as a determinant of poor prognosis in HCC and illustrated its impact on immune cell infiltration. After selecting arterial phase CT, we chose 7 whole-tumor features and 3 features covering both the tumor and its surroundings to create WT and WP models for FOXM1 prediction. The WT model showed strong predictive capabilities for FOXM1 expression by PR curve. Conversely, the WP model did not demonstrate the good predictive ability. In our study, the radiomics score (RS) was derived from whole-tumor regions on CT images. The RS was significantly associated with FOXM1 expression, with an AUC of 0.918 in the training cohort and 0.837 in the validation cohort. Furthermore, the RS was correlated with oxidative stress genes and was integrated with clinical variables to develop a nomogram, which demonstrated good calibration and discrimination in predicting 12-, 36-, and 60-month survival probabilities. Additionally, bioinformatics analysis revealed FOXM1's potential role in shaping the immune microenvironment, with its expression linked to immune cell infiltration. Conclusion This study highlights the potential of integrating FOXM1 expression and radiomics in understanding HCC's complexity. Our approach offers a new perspective in utilizing radiomics for non-invasive tumor characterization and suggests its potential in providing insights into molecular profiles. Further research is needed to validate these findings and explore their clinical implications in HCC management.
Collapse
Affiliation(s)
- Xianyu Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yongsheng Tang
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Donghao Wu
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ruixi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhiqun Lin
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xuhui Zhou
- Department of Radiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hezhen Wang
- Department of Radiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hang Zhai
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Junming Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xianjie Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Guangquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
14
|
Prazanowska KH, Lim SB. An integrated single-cell transcriptomic dataset for non-small cell lung cancer. Sci Data 2023; 10:167. [PMID: 36973297 PMCID: PMC10042991 DOI: 10.1038/s41597-023-02074-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
As single-cell RNA sequencing (scRNA-seq) has emerged as a great tool for studying cellular heterogeneity within the past decade, the number of available scRNA-seq datasets also rapidly increased. However, reuse of such data is often problematic due to a small cohort size, limited cell types, and insufficient information on cell type classification. Here, we present a large integrated scRNA-seq dataset containing 224,611 cells from human primary non-small cell lung cancer (NSCLC) tumors. Using publicly available resources, we pre-processed and integrated seven independent scRNA-seq datasets using an anchor-based approach, with five datasets utilized as reference and the remaining two, as validation. We created two levels of annotation based on cell type-specific markers conserved across the datasets. To demonstrate usability of the integrated dataset, we created annotation predictions for the two validation datasets using our integrated reference. Additionally, we conducted a trajectory analysis on subsets of T cells and lung cancer cells. This integrated data may serve as a resource for studying NSCLC transcriptome at the single cell level.
Collapse
Affiliation(s)
- Karolina Hanna Prazanowska
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea.
| |
Collapse
|
15
|
Fetisov TI, Borunova AA, Antipova AS, Antoshina EE, Trukhanova LS, Gorkova TG, Zuevskaya SN, Maslov A, Gurova K, Gudkov A, Lesovaya EA, Belitsky GA, Yakubovskaya MG, Kirsanov KI. Targeting Features of Curaxin CBL0137 on Hematological Malignancies In Vitro and In Vivo. Biomedicines 2023; 11:biomedicines11010230. [PMID: 36672738 PMCID: PMC9856019 DOI: 10.3390/biomedicines11010230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/31/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
The anticancer activity of Curaxin CBL0137, a DNA-binding small molecule with chromatin remodulating effect, has been demonstrated in different cancers. Herein, a comparative evaluation of CBL0137 activity was performed in respect to acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia and multiple myeloma (MM) cultured in vitro. MTT assay showed AML and MM higher sensitivity to CBL0137's cytostatic effect comparatively to other hematological malignancy cells. Flow cytometry cell cycle analysis revealed an increase in subG1 and G2/M populations after CBL0137 cell treatment, but the prevalent type of arrest varied. Apoptosis activation by CBL0137 measured by Annexin-V/PI dual staining was more active in AML and MM cells. RT2 PCR array showed that changes caused by CBL0137 in signaling pathways involved in cancer pathogenesis were more intensive in AML and MM cells. On the murine model of AML WEHI-3, CBL0137 showed significant anticancer effects in vivo, which were evaluated by corresponding changes in spleen and liver. Thus, more pronounced anticancer effects of CBL0137 in vitro were observed in respect to AML and MM. Experiments in vivo also indicated the perspective of CBL0137 use for AML treatment. This in accordance with the frontline treatment approach in AML using epigenetic drugs.
Collapse
Affiliation(s)
- Timur I. Fetisov
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Anna A. Borunova
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Alina S. Antipova
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Elena E. Antoshina
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Lubov S. Trukhanova
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Tatyana G. Gorkova
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | | | - Alexei Maslov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Katerina Gurova
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Andrei Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Ekaterina A. Lesovaya
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
- Department of Oncology, I.P. Pavlov Ryazan State Medical University, 390026 Ryazan, Russia
| | - Gennady A. Belitsky
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | | | - Kirill I. Kirsanov
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
- Correspondence:
| |
Collapse
|
16
|
Cancer secretome: finding out hidden messages in extracellular secretions. Clin Transl Oncol 2022; 25:1145-1155. [PMID: 36525229 DOI: 10.1007/s12094-022-03027-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
Secretome analysis has gained popularity recently as a very well-designed proteomic approach that is being used to study various interactions and their effects on cellular activity. This analysis is especially helpful while studying the effects of the cells on their microenvironment, paracrine and autocrine processes, their therapeutic purposes, and as a new diagnostic perspective. Cancer is a condition rather than a specific type of disease and is still yet to be fully understood. Cancer secretome is a fairly new concept that is being implemented to examine the interactions taking place in the tumor microenvironment and can help to understand the phenomena like induction of tumorigenesis, stimulation of immune cells, etc. The secretome analysis helps to gain a different perspective on the existing knowledge on cancer and its effects. The recent advances in secretome studies are directed toward secreted components as drug targets, biomarkers, and companion tools for diagnostic and prognostic purposes in cancer. This review aims to find the interactors in different types of cancer and understand the existing unstructured secretome data and its application in prognosis, diagnosis, and in biomarker study.
Collapse
|
17
|
Szeliga M, Rola R. Menadione Potentiates Auranofin-Induced Glioblastoma Cell Death. Int J Mol Sci 2022; 23:ijms232415712. [PMID: 36555352 PMCID: PMC9778806 DOI: 10.3390/ijms232415712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor. Recently, agents increasing the level of oxidative stress have been proposed as anticancer drugs. However, their efficacy may be lowered by the cytoprotective activity of antioxidant enzymes, often upregulated in neoplastic cells. Here, we assessed the mRNA and protein expression of thioredoxin reductase 1 (TrxR1), a master regulator of cellular redox homeostasis, in GBM and non-tumor brain tissues. Next, we examined the influence of an inhibitor of TrxR1, auranofin (AF), alone or in combination with a prooxidant menadione (MEN), on growth of GBM cell lines, patient-derived GBM cells and normal human astrocytes. We detected considerable amount of TrxR1 in the majority of GBM tissues. Treatment with AF decreased viability of GBM cells and their potential to form colonies and neurospheres. Moreover, it increased the intracellular level of reactive oxygen species (ROS). Pre-treatment with ROS scavenger prevented the AF-induced cell death, pointing to the important role of ROS in the reduction of cell viability. The cytotoxic effect of AF was potentiated by treatment with MEN. In conclusion, our results identify TrxR1 as an attractive drug target and highlights AF as an off-patent drug candidate in GBM therapy.
Collapse
Affiliation(s)
- Monika Szeliga
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland
- Correspondence: ; Tel.: +48-226086416
| | - Radosław Rola
- Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, 8 Jaczewski-ego Str., 20-090 Lublin, Poland
| |
Collapse
|
18
|
Yue Y, Tan M, Luo Y, Deng P, Wang H, Li J, Hao R, Tian L, Xie J, Chen M, Yu Z, Zhou Z, Pi H. miR-3614-5p downregulation promotes cadmium-induced breast cancer cell proliferation and metastasis by targeting TXNRD1. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114270. [PMID: 36335879 DOI: 10.1016/j.ecoenv.2022.114270] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
Cadmium (Cd), which is considered an endocrine disruptor, has been linked to the onset of breast cancer (BC). Our recent study demonstrated that Cd-induced BC progression has a strong correlation with miR-374c-5p dysregulation. The aim of our work was to investigate other potential miRNAs involved in Cd-induced BC cell proliferation and metastasis. In our study, the miRNA profiles of Cd-treated T-47D cells (10 μM, 72 h) were analyzed by miRNA-seq, and our results confirmed that miR-3614-5p was the top downregulated miRNA. Moreover, miR-3614-5p mimic transfection significantly decreased the proliferative ability, migration and invasive ability of BC cell lines (T-47D and MCF-7). Furthermore, we analyzed the overlapping genes from our RNA-seq data and predicted targets from the mirDIP database, and twelve genes (ALDH1A3, FBN1, GRIA3, NOS1, PLD5, PTGER4, RASGRF2, RELN, RNF150, SLC17A4, TG, and TXNRD1) were identified as potential binding targets of miR-3614-5p in the current model. Nonetheless, only miR-3614-5p inhibition caused an increase in TXNRD1 expression upon Cd exposure in T-47D and MCF-7 cell lines. Importantly, luciferase reporter assays further verified that miR-3614-5p suppressed the expression of TXNRD1 by directly binding to the 3'-untranslated region (UTR), and TXNRD1 inhibition significantly repressed the proliferation and metastasis capacity of BC cells upon Cd exposure. Together, our findings demonstrated that Cd exposure repressed the expression of miR-3614-5p, thus activating TXNRD1 expression, which promoted the abnormal proliferation and metastasis of BC cells.
Collapse
Affiliation(s)
- Yang Yue
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Miduo Tan
- Surgery Department of Galactophore, Zhuzhou Hospital Affiliated to Xiangya Shool of Medicine, Central South University, Zhuzhou 412000, Hunan, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Hui Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Jingdian Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Rongrong Hao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
19
|
Qi X, Wan Z, Jiang B, Ouyang Y, Feng W, Zhu H, Tan Y, He R, Xie L, Li Y. Inducing ferroptosis has the potential to overcome therapy resistance in breast cancer. Front Immunol 2022; 13:1038225. [PMID: 36505465 PMCID: PMC9730886 DOI: 10.3389/fimmu.2022.1038225] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is the most common type of malignancy among women. Due to the iron-dependent character of breast cancer cells, they are more sensitive to ferroptosis compared to normal cells. It is possible to reverse tumor resistance by inducing ferroptosis in breast cancer cells, thereby improving tumor treatment outcomes. Ferroptosis is highly dependent on the balance of oxidative and antioxidant status. When ferroptosis occurs, intracellular iron levels are significantly increased, leading to increased membrane lipid peroxidation and ultimately triggering ferroptosis. Ferroptotic death is a form of autophagy-associated cell death. Synergistic use of nanoparticle-loaded ferroptosis-inducer with radiotherapy and chemotherapy achieves more significant tumor suppression and inhibits the growth of breast cancer by targeting cancer tissues, enhancing the sensitivity of cells to drugs, reducing the drug resistance of cancer cells and the toxicity of drugs. In this review, we present the current status of breast cancer and the mechanisms of ferroptosis. It is hopeful for us to realize effective treatment of breast cancer through targeted ferroptosis.
Collapse
Affiliation(s)
- Xiaowen Qi
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhixing Wan
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Baohong Jiang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuhan Ouyang
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjie Feng
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hongbo Zhu
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yeru Tan
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Rongfang He
- Department of Pathology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liming Xie
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuehua Li
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
20
|
Al Khamici H, Sanchez VC, Yan H, Cataisson C, Michalowski AM, Yang HH, Li L, Lee MP, Huang J, Yuspa SH. The oxidoreductase CLIC4 is required to maintain mitochondrial function and resistance to exogenous oxidants in breast cancer cells. J Biol Chem 2022; 298:102275. [PMID: 35863434 PMCID: PMC9418444 DOI: 10.1016/j.jbc.2022.102275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
The chloride intracellular channel-4 (CLIC4) is one of the six highly conserved proteins in the CLIC family that share high structural homology with GST-omega in the GST superfamily. While CLIC4 is a multifunctional protein that resides in multiple cellular compartments, the discovery of its enzymatic glutaredoxin-like activity in vitro suggested that it could function as an antioxidant. Here, we found that deleting CLIC4 from murine 6DT1 breast tumor cells using CRISPR enhanced the accumulation of reactive oxygen species (ROS) and sensitized cells to apoptosis in response to H2O2 as a ROS-inducing agent. In intact cells, H2O2 increased the expression of both CLIC4 mRNA and protein. In addition, increased superoxide production in 6DT1 cells lacking CLIC4 was associated with mitochondrial hyperactivity including increased mitochondrial membrane potential and mitochondrial organelle enlargement. In the absence of CLIC4, however, H2O2-induced apoptosis was associated with low expression and degradation of the antiapoptotic mitochondrial protein Bcl2 and the negative regulator of mitochondrial ROS, UCP2. Furthermore, transcriptomic profiling of H2O2-treated control and CLIC4-null cells revealed upregulation of genes associated with ROS-induced apoptosis and downregulation of genes that sustain mitochondrial functions. Accordingly, tumors that formed from transplantation of CLIC4-deficient 6DT1 cells were highly necrotic. These results highlight a critical role for CLIC4 in maintaining redox-homeostasis and mitochondrial functions in 6DT1 cells. Our findings also raise the possibility of targeting CLIC4 to increase cancer cell sensitivity to chemotherapeutic drugs that are based on elevating ROS in cancer cells.
Collapse
Affiliation(s)
- Heba Al Khamici
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Vanesa C Sanchez
- Office of Science, Division of Nonclinical Science, Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hualong Yan
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Aleksandra M Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Luowei Li
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Jing Huang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA.
| |
Collapse
|
21
|
Li Q, Jiang Y, Song N, Zhou B, Li Z, Lin L. An Immune-Related Genetic Feature Depicted the Heterogeneous Nature of Lung Adenocarcinoma and Squamous Cell Carcinoma and Their Distinctive Predicted Drug Responses. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8447083. [PMID: 36071867 PMCID: PMC9442502 DOI: 10.1155/2022/8447083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
One of the primary causes of global cancer-associated mortality is lung cancer (LC). Current improvements in the management of LC rely mainly on the advancement of patient stratification, both molecularly and clinically, to achieve the maximal therapeutic benefit, while most LC screening protocols remain underdeveloped. In this research, we first employed two algorithms (ESTIMATE and xCell) to calculate the immune/stromal infiltration scores. This helped identify the altered immune infiltration landscapes in lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC). Afterward, based on their immune-related characteristics, we successfully stratified the LUAD and LUSC into 2 and 3 clusters, respectively. Different from the conventional bioinformatic approaches that start from the investigation of differential expression of single genes, differentially enriched curated gene sets identified through gene set variation analyses (GSVA) were curated, and gene names were reconstructed afterward. Furthermore, weighted gene correlation network analyses (WGCNA) were used to reveal hub genes highly connected with the clustering process. Actual expression levels of critical hub genes among different clusters were compared and so were the functional pathways these genes enriched into. Lastly, a computational method was applied to predict and compare the responses of each cluster to primary therapeutic agents. The heterogeneity presented in our study, along with the drug responses expected for identified clusters, may shed light on future exploration of combination immunochemotherapy that facilitates the optimization of individualized therapy.
Collapse
Affiliation(s)
- Qiuyuan Li
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Yan Jiang
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Nan Song
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Bin Zhou
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Zhao Li
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| | - Lei Lin
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, No. 507 Zhengmin Rd., Shanghai, China
| |
Collapse
|
22
|
Tian Y, Ge Z, Xu M, Ge X, Zhao M, Ding F, Yin J, Wang X, You Y, Shi Z, Qian X. Diallyl trisulfide sensitizes radiation therapy on glioblastoma through directly targeting thioredoxin 1. Free Radic Biol Med 2022; 189:157-168. [PMID: 35921994 DOI: 10.1016/j.freeradbiomed.2022.07.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Radiotherapy is a standard-of-care treatment approach for glioblastoma (GBM) patients, but therapeutic resistance to radiotherapy remains a major challenge. Here we demonstrate that diallyl trisulfide (DATS) directly conjugates with cysteine (C) 32 and C35 (C32/35) residues of thioredoxin 1 (Trx1) through Michael addition reactions. Due to localizing in activity center of Trx1, the conjugation between DATS and C32/35 results in inhibition of Trx1 activity, therefore disturbing thioredoxin system and leading to accumulated levels of reactive oxygen species (ROS). High levels of Trx1 expression are correlated with poor prognosis of glioma patients. Notably, we reveal that DATS synergistically enhances irradiation (IR)-induced ROS accumulation, apoptosis, DNA damage, as well as inhibition of tumor growth of GBM cells. These findings highlight the potential benefits of DATS in sensitizing radiotherapy of GBM patients.
Collapse
Affiliation(s)
- Yangyang Tian
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zehe Ge
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Miao Xu
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Ge
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Mengjie Zhao
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Fangshu Ding
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianxing Yin
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xiuxing Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; National Health Commission Key Laboratory of Antibody Technologies, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yongping You
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhumei Shi
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Xu Qian
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
23
|
Systematic Oxidative Stress Indexes Associated with the Prognosis in Patients with T Lymphoblastic Lymphoma/Leukemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2679154. [PMID: 35965687 PMCID: PMC9371838 DOI: 10.1155/2022/2679154] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/23/2022] [Indexed: 11/22/2022]
Abstract
Background T lymphoblastic lymphoma/leukemia (T-LBL/ALL) is an aggressive malignant tumor with 5-year overall survival (OS) rate reached 80% after high-dose chemotherapy. Due to the relatively low incidence of T-LBL/ALL, only a few risk factors have been identified. The occurrence and prognosis of malignant tumors are closely related to oxidative stress, but the prognostic relationship between T-LBL/ALL and systematic oxidative stress indexes has not been reported yet. Methods A total of 258 T-LBL/ALL patients were retrospectively analyzed. The relationship between systematic oxidative stress indexes, such as creatinine (CRE), gamma-glutamyl transpeptidase (γ-GGT), albumin (ALB), alkaline phosphatase (ALP), fibrinogen (FBG), C-reactive protein (CRP) and total bilirubin (TBIL), and survival of T-LBL/ALL patients, was analyzed. The weight of indexes was used to calculate the patients' oxidative stress risk score. The independent prognostic value of oxidative stress risk grouping (OSRG) was analyzed. Results Higher CRE, CRP, and lower ALB were associated with poorer OS in T-LBL/ALL patients. The OSRG established by CRE, ALB, and CRP was an independent prognostic factor for OS of T-LBL/ALL patients. Patients in the high-risk group were more likely to be patients older than 14 years old and patients with superior vena cava obstruction syndrome (SVCS), pleural effusion, pericardial effusion, and mediastinal mass. Conclusion For OS in T-LBL/ALL patients, OSRG was observed as an independent prognostic factor, which provided a new idea for accurate prognostic prediction.
Collapse
|
24
|
Mukherjee S, Dutta A, Chakraborty A. The interaction of oxidative stress with MAPK, PI3/AKT, NF-κB, and DNA damage kinases influences the fate of γ-radiation-induced bystander cells. Arch Biochem Biophys 2022; 725:109302. [DOI: 10.1016/j.abb.2022.109302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/27/2022] [Accepted: 05/22/2022] [Indexed: 11/02/2022]
|
25
|
Vitamin D 3 and Salinomycin synergy in MCF-7 cells cause cell death via endoplasmic reticulum stress in monolayer and 3D cell culture. Toxicol Appl Pharmacol 2022; 452:116178. [PMID: 35914560 DOI: 10.1016/j.taap.2022.116178] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 11/21/2022]
Abstract
1α, 25, dihydroxyvitamin D3 (1,25D), the active form of vitamin D3, has antitumor properties in several cancer cell lines in vitro. Salinomycin (Sal) has anticancer activity against cancer cell lines. This study aims to examine the cytotoxic and antiproliferative effect of Sal associated with 1,25D on MCF-7 breast carcinoma cell line cultured in monolayer (2D) and three-dimensional models (mammospheres). We also aim to evaluate the molecular mechanism of Sal and 1,25D-mediated effects. We report that Sal and 1,25D act synergistically in MCF-7 mammospheres and monolayer causing G1 cell cycle arrest, reduction of mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) overproduction with a long-lasting cytotoxic response represented by clonogenic and mammosphere assay. We observed the induction of cell death by apoptosis with upregulation in mRNA levels of apoptosis-related genes (CASP7, CASP9, and BBC3). Extensive cytoplasmic vacuolization, a morphological characteristic found in paraptosis, was also seen and could be triggered by endoplasmic reticulum stress (ER) as we found transcriptional upregulation of genes related to ER stress (ATF6, GADD153, GADD45G, EIF2AK3, and HSPA5). Overall, Sal and 1,25D act synergistically, inhibiting cell proliferation by activating simultaneously multiple death pathways and may be a novel and promising luminal A breast cancer therapy strategy.
Collapse
|
26
|
Manuck TA, Eaves LA, Rager JE, Sheffield-abdullah K, Fry RC. Nitric oxide-related gene and microRNA expression in peripheral blood in pregnancy vary by self-reported race. Epigenetics 2022; 17:731-745. [PMID: 34308756 PMCID: PMC9336489 DOI: 10.1080/15592294.2021.1957576] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Adverse pregnancy outcomes disproportionately affect non-Hispanic (NH) Black patients in the United States. Structural racism has been associated with increased psychosocial distress and inflammation and may trigger oxidative stress. Thus, the nitric oxide (NO) pathway (involved in the regulation of inflammation and oxidative stress) may partly explain the underlying disparities in obstetric outcomes.Cohort study of 154 pregnant patients with high-risk obstetric histories; n = 212 mRNAs and n = 108 microRNAs (miRNAs) in the NO pathway were evaluated in circulating white blood cells. NO pathway mRNA and miRNA transcript counts were compared by self-reported race; NH Black patients were compared with women of other races/ethnicities. Finally, miRNA-mRNA expression levels were correlated.Twenty-two genes (q < 0.10) were differentially expressed in self-identified NH Black individuals. Superoxide dismutase 1 (SOD1), interleukin-8 (IL-8), dynein light chain LC8-type 1 (DYNLL1), glutathione peroxidase 4 (GPX4), and glutathione peroxidase 1 (GPX1) were the five most differentially expressed genes among NH Black patients compared to other patients. There were 63 significantly correlated miRNA-mRNA pairs (q < 0.10) demonstrating potential miRNA regulation of associated target mRNA expression. Ten miRNAs that were identified as members of significant miRNA-mRNA pairs were also differentially expressed among NH Black patients (q < 0.10).These findings support an association between NO pathway and inflammation and infection-related mRNA and miRNA expression in blood drawn during pregnancy and patient race/ethnicity. These findings may reflect key differences in the biology of inflammatory gene dysregulation that occurs in response to the stress of systemic racism and that underlies disparities in pregnancy outcomes.
Collapse
Affiliation(s)
- Tracy A. Manuck
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, United States
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Lauren A. Eaves
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Julia E Rager
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | | | - Rebecca C. Fry
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| |
Collapse
|
27
|
Zhang J, Chen Y, Fang J. Targeting thioredoxin reductase by micheliolide contributes to radiosensitizing and inducing apoptosis of HeLa cells. Free Radic Biol Med 2022; 186:99-109. [PMID: 35561844 DOI: 10.1016/j.freeradbiomed.2022.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 12/30/2022]
Abstract
Inhibition of thioredoxin reductase (TrxR) is a crucial strategy for the discovery of antineoplastic drugs and radiosensitizers. As an anticancer candidate derived from Michelia, micheliolide (MCL) is converted readily from parthenolide (PTL), and has better stability and solubility than PTL. However, the anticancer mechanism of MCL has not been fully dissected. We present here for the first time that MCL-targeted inhibition of TrxR not only promotes oxidative stress-mediated HeLa cell apoptosis but also sensitizes ionizing radiation (IR) treatment. Further mechanistic studies demonstrate that MCL covalently binds to Sec at position 498 of TrxR to restrain the biological function of TrxR. It exhibits the inhibition of TrxR activity, enhancement of oxidized Trx, and sensitization of IR in the cellular environment, accompanied by the accumulation of reactive oxygen species (ROS) and the collapse of the intracellular redox balance. In addition, HeLa-shTrxR1 cells with knockdown of TrxR were more sensitive than the HeLa-shNT cells to either MCL-treated or IR-induced cytotoxicity, ROS, and apoptosis, suggesting that inhibition of TrxR by MCL is likely responsible for increased cytotoxicity and enhanced radiation response. These findings further establish the mechanistic understanding and preclinical data to support the further investigation of MCL's potential as a prospective radiosensitizer and cancer chemotherapeutic agent.
Collapse
Affiliation(s)
- Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, And College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Yaxiong Chen
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, And College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, And Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Jianguo Fang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, And College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, 210094, China.
| |
Collapse
|
28
|
Xu T, Jiang Y, Yuan S, Zhang L, Chen X, Zhao W, Cai L, Xiao B, Jia L. Andrographolide Inhibits ER-Positive Breast Cancer Growth and Enhances Fulvestrant Efficacy via ROS-FOXM1-ER-α Axis. Front Oncol 2022; 12:899402. [PMID: 35615146 PMCID: PMC9124841 DOI: 10.3389/fonc.2022.899402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Estrogen receptor (ER)-positive breast cancer is the main subtype of breast cancer (BRCA) with high incidence and mortality. Andrographolide (AD), a major active component derived from the traditional Chinese medicine Andrographis paniculate, has substantial anti-cancer effect in various tumors. However, the antitumor efficacy and the underlying molecular mechanisms of AD on ER-positive breast cancer are poorly understood. In the present study, we demonstrated that andrographolide (AD) significantly inhibited the growth of ER-positive breast cancer cells. Mechanistically, AD suppressed estrogen receptor 1 (ESR1, encodes ER-α) transcription to inhibit tumor growth. Further studies revealed that AD induced ROS production to down-regulate FOXM1-ER-α axis. Conversely, inhibiting ROS production with N-acetylcysteine (NAC) elevated AD-decreased ER-α expression, which could be alleviated by FOXM1 knockdown. In addition, AD in combination with fulvestrant (FUL) synergistically down-regulated ER-α expression to inhibit ER-positive breast cancer both in vitro and in vivo. These findings collectively indicate that AD suppresses ESR1 transcription through ROS-FOXM1 axis to inhibit ER-positive breast cancer growth and suggest that AD might be a potential therapeutic agent and fulvestrant sensitizer for ER-positive breast cancer treatment.
Collapse
|
29
|
Germline polymorphisms in genes involved in the antioxidant system predict the efficacy of cetuximab in metastatic colorectal cancer patients enrolled in FIRE-3 trial. Clin Colorectal Cancer 2022; 21:259-266. [DOI: 10.1016/j.clcc.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/14/2022] [Accepted: 05/19/2022] [Indexed: 11/21/2022]
|
30
|
Basak T, Kanwar RK. Iron imbalance in cancer: Intersection of deficiency and overload. Cancer Med 2022; 11:3837-3853. [PMID: 35460205 PMCID: PMC9582687 DOI: 10.1002/cam4.4761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/09/2022] [Accepted: 02/25/2022] [Indexed: 12/19/2022] Open
Abstract
Iron, an essential trace element, plays a complex role in tumour biology. While iron causes cancer clearance through toxic free radical generation, iron‐induced free radical flux also acts as a cancer promoter. These fates majorly guided through cellular response towards pro‐oxidant and antioxidant settings in a tumour microenvironment, designate iron‐induced oxidative stress as a common yet paradoxical factor in pro‐tumorigenesis as well as anti‐tumorigenesis, posing a challenge to laying down iron thresholds favouring tumour clearance. Additionally, complexity of iron's association with carcinogenesis has been extended to iron‐induced ROS's involvement in states of both iron deficiency and overload, conditions identified as comparable, inevitable and significant coexisting contributors as well as outcomes in chronic infections and tumorigenesis. Besides, iron overload may also develop as an unwanted outcome in certain cancer patients, as a result of symptomatic anaemia treatment owed to irrational iron‐restoration therapies without a prior knowledge of body's iron status with both conditions synergistically acting towards tumour aggravation. The co‐play of iron deficiency and overload along with iron's pro‐tumour and antitumour roles with intersecting mechanisms, thus presents an unpredictable regulatory response loop in a state of malignancy. The relevance of iron's thresholds beyond which it proves to be beneficial against tumorigenesis hence becomes questionable. These factors pose a challenge, over establishing if iron chelation or iron flooding acts as a better approach towards antitumour therapies. This review presents a critical picture of multiple contrasting features of iron's behaviour in cancer, leading towards two conditions lying at opposite ends of a spectrum: iron deficiency and overload in chronic disease conditions including cancer, hence, validating the critical significance of diagnosis of patients' iron status prior to opting for subsequent therapies.
Collapse
Affiliation(s)
- Tulika Basak
- Institute for Innovation in Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Faculty of Health, Deakin University, Geelong, Victoria, Australia
| | - Rupinder Kaur Kanwar
- Institute for Innovation in Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Faculty of Health, Deakin University, Geelong, Victoria, Australia.,Department of Translational Medicine Centre, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, India
| |
Collapse
|
31
|
Roca MS, Moccia T, Iannelli F, Testa C, Vitagliano C, Minopoli M, Camerlingo R, De Riso G, De Cecio R, Bruzzese F, Conte M, Altucci L, Di Gennaro E, Avallone A, Leone A, Budillon A. HDAC class I inhibitor domatinostat sensitizes pancreatic cancer to chemotherapy by targeting cancer stem cell compartment via FOXM1 modulation. J Exp Clin Cancer Res 2022; 41:83. [PMID: 35241126 PMCID: PMC8892808 DOI: 10.1186/s13046-022-02295-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/19/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) represents an unmet clinical need due to the very poor prognosis and the lack of effective therapy. Here we investigated the potential of domatinostat (4SC-202), a new class I histone deacetylase (HDAC) inhibitor, currently in clinical development, to sensitize PDAC to first line standard gemcitabine (G)/taxol (T) doublet chemotherapy treatment. METHODS Synergistic anti-tumor effect of the combined treatment was assessed in PANC1, ASPC1 and PANC28 PDAC cell lines in vitro as well as on tumor spheroids and microtissues, by evaluating combination index (CI), apoptosis, clonogenic capability. The data were confirmed in vivo xenograft models of PANC28 and PANC1 cells in athymic mice. Cancer stem cells (CSC) targeting was studied by mRNA and protein expression of CSC markers, by limiting dilution assay, and by flow cytometric and immunofluorescent evaluation of CSC mitochondrial and cellular oxidative stress. Mechanistic role of forkhead box M1 (FOXM1) and downstream targets was evaluated in FOXM1-overexpressing PDAC cells. RESULTS We showed that domatinostat sensitized in vitro and in vivo models of PDAC to chemotherapeutics commonly used in PDAC patients management and particularly to GT doublet, by targeting CSC compartment through the induction of mitochondrial and cellular oxidative stress. Mechanistically, we showed that domatinostat hampers the expression and function of FOXM1, a transcription factor playing a crucial role in stemness, oxidative stress modulation and DNA repair. Domatinostat reduced FOXM1 protein levels by downregulating mRNA expression and inducing proteasome-mediated protein degradation thus preventing nuclear translocation correlated with a reduction of FOXM1 target genes. Furthermore, by overexpressing FOXM1 in PDAC cells we significantly reduced domatinostat-inducing oxidative mitochondrial and cellular stress and abolished GT sensitization, both in adherent and spheroid cells, confirming FOXM1 crucial role in the mechanisms described. Finally, we found a correlation of FOXM1 expression with poor progression free survival in PDAC chemotherapy-treated patients. CONCLUSIONS Overall, we suggest a novel therapeutic strategy based on domatinostat to improve efficacy and to overcome resistance of commonly used chemotherapeutics in PDAC that warrant further clinical evaluation.
Collapse
Affiliation(s)
- Maria Serena Roca
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Tania Moccia
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Federica Iannelli
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Cristina Testa
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Michele Minopoli
- Neoplastic Progression Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Rosa Camerlingo
- Cell Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Giulia De Riso
- Department of molecular medicine and medical biotechnology, University of Naples "Federico II", Naples, Italy
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Francesca Bruzzese
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy.,BIOGEM, (AV), Naples, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy.
| |
Collapse
|
32
|
Lee YJ, Kim SW, Jung MH, Kim YS, Kim KS, Suh DS, Kim KH, Choi EH, Kim J, Kwon BS. Plasma-activated medium inhibits cancer stem cell-like properties and exhibits a synergistic effect in combination with cisplatin in ovarian cancer. Free Radic Biol Med 2022; 182:276-288. [PMID: 35276382 DOI: 10.1016/j.freeradbiomed.2022.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022]
Abstract
Ovarian cancer stem-like cells (CSCs) have been implicated in tumor recurrence, metastasis, and drug resistance. Accumulating evidence has demonstrated the antitumor effect of plasma-activated medium (PAM) in various carcinomas, including ovarian cancer. Thus, PAM represents a novel onco-therapeutic strategy. However, its impact on ovarian CSCs is unclear. Here, we show that ovarian CSCs resistant to high-dose conventional chemotherapeutic agents used for ovarian cancer treatment exhibited dose-dependent sensitivity to PAM. In addition, PAM treatment reduced the expression of stem cell markers and sphere formation, along with the aldehyde dehydrogenase- or CD133-positive cell population. We further investigated the effect of PAM in combination with other chemotherapeutics on ovarian CSCs in vitro. PAM exhibited synergistic cytotoxicity with cisplatin (CDDP) but not with paclitaxel and doxorubicin. In a peritoneal metastasis xenograft model established via intraperitoneal spheroid injection, PAM intraperitoneal therapy significantly suppressed peritoneal carcinomatosis (tumor size and number), with a more significant decrease observed due to the combined effects of PAM and CDDP with no side effects. Taken together, our results indicate that PAM inhibits ovarian CSC traits and exhibits synergetic cytotoxicity with CDDP, demonstrating PAM as a promising intraparietal chemotherapy for enhancing antitumor efficacy and reducing side effects.
Collapse
Affiliation(s)
- Young Joo Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul, 02447, South Korea
| | - Sung Wook Kim
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul, 02447, South Korea
| | - Min Hyung Jung
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul, 02447, South Korea
| | - Young Sun Kim
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul, 02447, South Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Dong Soo Suh
- Departments of Obstetrics and Gynecology, Medical Research Institute, Pusan National University School of Medicine, Busan, South Korea
| | - Ki Hyung Kim
- Departments of Obstetrics and Gynecology, Medical Research Institute, Pusan National University School of Medicine, Busan, South Korea
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Kwangwoon University, Seoul, 139-701, South Korea
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women's University, Seoul, 04310, South Korea; Research Institute for Women's Health, Sookmyung Women's University, Seoul, 04310, South Korea.
| | - Byung Su Kwon
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul, 02447, South Korea.
| |
Collapse
|
33
|
AKR1B1 as a Prognostic Biomarker of High-Grade Serous Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14030809. [PMID: 35159076 PMCID: PMC8834204 DOI: 10.3390/cancers14030809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary We evaluated the levels of AKR1B1 and AKR1B10 in 99 patients with high-grade serous ovarian cancer and their association with clinicopathological characteristics, survival, and response to chemotherapy. An immunohistochemical analysis showed that higher AKR1B1 levels correlated with a better disease-free survival of patients whereas we saw no differences for AKR1B10 levels. A multivariant Cox analysis identified high AKR1B1 levels as an important prognostic factor for both overall and disease-free survival. A further analysis revealed no association between AKR1B1 and AKR1B10 levels and response to chemotherapy. Abstract Although aldo-keto reductases (AKRs) have been widely studied in cancer, no study to date has examined the roles of AKR family 1 members B1 (AKR1B1) and B10 (AKR1B10) in a large group of ovarian cancer patients. AKR1B1 and AKR1B10 play a significant role in inflammation and the metabolism of different chemotherapeutics as well as cell differentiation, proliferation, and apoptosis. Due to these functions, we examined the potential of AKR1B1 and AKR1B10 as tissue biomarkers. We assessed the immunohistochemical levels of AKR1B1 and AKR1B10 in tissue paraffin sections from 99 patients with high-grade serous ovarian cancer (HGSC) and compared these levels with clinicopathological characteristics, survival, and response to chemotherapy. A higher immunohistochemical AKR1B1 expression correlated with a better overall and disease-free survival of HGSC patients whereas AKR1B10 expression did not show any significant differences. A multivariant Cox analysis demonstrated that a high AKR1B1 expression was an important prognostic factor for both overall and disease-free survival. However, AKR1B1 and AKR1B10 were not associated with different responses to chemotherapy. Our data suggest that AKR1B1 is involved in the pathogenesis of HGSC and is a potential prognostic biomarker for this cancer.
Collapse
|
34
|
An Integrated In Silico, In Vitro and Tumor Tissues Study Identified Selenoprotein S (SELENOS) and Valosin-Containing Protein (VCP/p97) as Novel Potential Associated Prognostic Biomarkers in Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14030646. [PMID: 35158912 PMCID: PMC8833666 DOI: 10.3390/cancers14030646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Triple negative breast cancer (TNBC) represents a clinical challenge because its early relapse, poor overall survival and lack of effective treatments. Altered levels selenoproteins have been correlated with development and progression of some cancers, however, no consistent data are available about their involvement in TNBC. Here we analyzed the expression profile of all twenty-five human selenoproteins in TNBC cells and tissues by a systematic approach, integrating in silico and wet lab approaches. We showed that the expression profiles of five selenoproteins are specifically dysregulated in TNBC. Most importantly, by a bioinformatics analysis, we selected selenoprotein S and its interacting protein valosin-containing protein (VCP/p97) as inter-related with the others and whose coordinated over-expression is associated with poor prognosis in TNBC. Overall, we highlighted two mechanistically related novel proteins whose correlated expression could be exploited for a better definition of prognosis as well as suggested as novel therapeutic target in TNBC. Abstract Background. Triple negative breast cancer (TNBC) is a heterogeneous group of tumors with early relapse, poor overall survival, and lack of effective treatments. Hence, new prognostic biomarkers and therapeutic targets are needed. Methods. The expression profile of all twenty-five human selenoproteins was analyzed in TNBC by a systematic approach.In silicoanalysis was performed on publicly available mRNA expression datasets (Cancer Cell Line Encyclopedia, CCLE and Library of Integrated Network-based Cellular Signatures, LINCS). Reverse transcription quantitative PCR analysis evaluated selenoprotein mRNA expression in TNBC versus non-TNBC and normal breast cells, and in TNBC tissues versus normal counterparts. Immunohistochemistry was employed to study selenoproteins in TNBC tissues. STRING and Cytoscape tools were used for functional and network analysis. Results.GPX1, GPX4, SELENOS, TXNRD1 and TXNRD3 were specifically overexpressed in TNBC cells, tissues and CCLE/LINCS datasets. Network analysis demonstrated that SELENOS-binding valosin-containing protein (VCP/p97) played a critical hub role in the TNBCselenoproteins sub-network, being directly associated with SELENOS expression. The combined overexpression of SELENOS and VCP/p97 correlated with advanced stages and poor prognosis in TNBC tissues and the TCGA dataset. Conclusion. Combined evaluation of SELENOS and VCP/p97 might represent a novel potential prognostic signature and a therapeutic target to be exploited in TNBC.
Collapse
|
35
|
Mai Z, Chen H, Huang M, Zhao X, Cui L. A Robust Metabolic Enzyme-Based Prognostic Signature for Head and Neck Squamous Cell Carcinoma. Front Oncol 2022; 11:770241. [PMID: 35127477 PMCID: PMC8810637 DOI: 10.3389/fonc.2021.770241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is still a menace to public wellbeing globally. However, the underlying molecular events influencing the carcinogenesis and prognosis of HNSCC are poorly known. Methods Gene expression profiles of The Cancer Genome Atlas (TCGA) HNSCC dataset and GSE37991 were downloaded from the TCGA database and gene expression omnibus, respectively. The common differentially expressed metabolic enzymes (DEMEs) between HNSCC tissues and normal controls were screened out. Then a DEME-based molecular signature and a clinically practical nomogram model were constructed and validated. Results A total of 23 commonly upregulated and 9 commonly downregulated DEMEs were identified in TCGA HNSCC and GSE37991. Gene ontology analyses of the common DEMEs revealed that alpha-amino acid metabolic process, glycosyl compound metabolic process, and cellular amino acid metabolic process were enriched. Based on the TCGA HNSCC cohort, we have built up a robust DEME-based prognostic signature including HPRT1, PLOD2, ASNS, TXNRD1, CYP27B1, and FUT6 for predicting the clinical outcome of HNSCC. Furthermore, this prognosis signature was successfully validated in another independent cohort GSE65858. Moreover, a potent prognostic signature-based nomogram model was constructed to provide personalized therapeutic guidance for treating HNSCC. In vitro experiment revealed that the knockdown of TXNRD1 suppressed malignant activities of HNSCC cells. Conclusion Our study has successfully developed a robust DEME-based signature for predicting the prognosis of HNSCC. Moreover, the nomogram model might provide useful guidance for the precision treatment of HNSCC.
Collapse
Affiliation(s)
- Zizhao Mai
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Huan Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Mingshu Huang
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xinyuan Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Xinyuan Zhao, ; Li Cui,
| | - Li Cui
- Stomatological Hospital, Southern Medical University, Guangzhou, China
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA, United States
- *Correspondence: Xinyuan Zhao, ; Li Cui,
| |
Collapse
|
36
|
Alghamdi RA, Exposito-Rodriguez M, Mullineaux PM, Brooke GN, Laissue PP. Assessing Phototoxicity in a Mammalian Cell Line: How Low Levels of Blue Light Affect Motility in PC3 Cells. Front Cell Dev Biol 2021; 9:738786. [PMID: 34977004 PMCID: PMC8718804 DOI: 10.3389/fcell.2021.738786] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/16/2021] [Indexed: 11/24/2022] Open
Abstract
Phototoxicity is a significant constraint for live cell fluorescence microscopy. Excessive excitation light intensities change the homeostasis of the observed cells. Erroneous and misleading conclusions may be the problematic consequence of observing such light-induced pathophysiology. In this study, we assess the effect of blue light, as commonly used for GFP and YFP excitation, on a motile mammalian cell line. Tracking PC3 cells at different light doses and intensities, we show how motility can be used to reliably assess subtle positive and negative effects of illumination. We further show that the effects are a factor of intensity rather than light dose. Mitotic delay was not a sensitive indicator of phototoxicity. For early detection of the effect of blue light, we analysed the expression of genes involved in oxidative stress. This study addresses the need for relatively simple and sensitive methods to establish a dose-response curve for phototoxicity in mammalian cell line models. We conclude with a working model for phototoxicity and recommendations for its assessment.
Collapse
Affiliation(s)
- Rana A. Alghamdi
- Department of Chemistry, Science and Arts College, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia
- School of Life Sciences, University of Essex, Colchester, United Kingdom
| | - Marino Exposito-Rodriguez
- School of Life Sciences, University of Essex, Colchester, United Kingdom
- Sainsbury Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | | - Greg N. Brooke
- School of Life Sciences, University of Essex, Colchester, United Kingdom
| | - Philippe P. Laissue
- School of Life Sciences, University of Essex, Colchester, United Kingdom
- *Correspondence: Philippe P. Laissue,
| |
Collapse
|
37
|
Linke R, Limmer M, Juranek SA, Heine A, Paeschke K. The Relevance of G-Quadruplexes for DNA Repair. Int J Mol Sci 2021; 22:12599. [PMID: 34830478 PMCID: PMC8620898 DOI: 10.3390/ijms222212599] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/28/2023] Open
Abstract
DNA molecules can adopt a variety of alternative structures. Among these structures are G-quadruplex DNA structures (G4s), which support cellular function by affecting transcription, translation, and telomere maintenance. These structures can also induce genome instability by stalling replication, increasing DNA damage, and recombination events. G-quadruplex-driven genome instability is connected to tumorigenesis and other genetic disorders. In recent years, the connection between genome stability, DNA repair and G4 formation was further underlined by the identification of multiple DNA repair proteins and ligands which bind and stabilize said G4 structures to block specific DNA repair pathways. The relevance of G4s for different DNA repair pathways is complex and depends on the repair pathway itself. G4 structures can induce DNA damage and block efficient DNA repair, but they can also support the activity and function of certain repair pathways. In this review, we highlight the roles and consequences of G4 DNA structures for DNA repair initiation, processing, and the efficiency of various DNA repair pathways.
Collapse
Affiliation(s)
- Rebecca Linke
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127 Bonn, Germany; (R.L.); (M.L.); (S.A.J.); (A.H.)
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michaela Limmer
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127 Bonn, Germany; (R.L.); (M.L.); (S.A.J.); (A.H.)
| | - Stefan A. Juranek
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127 Bonn, Germany; (R.L.); (M.L.); (S.A.J.); (A.H.)
| | - Annkristin Heine
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127 Bonn, Germany; (R.L.); (M.L.); (S.A.J.); (A.H.)
| | - Katrin Paeschke
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127 Bonn, Germany; (R.L.); (M.L.); (S.A.J.); (A.H.)
| |
Collapse
|
38
|
Jin LY, Gu YL, Zhu Q, Li XH, Jiang GQ. The role of ferroptosis-related genes for overall survival prediction in breast cancer. J Clin Lab Anal 2021; 35:e24094. [PMID: 34741349 PMCID: PMC8649350 DOI: 10.1002/jcla.24094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/11/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
Background Ferroptosis is a novel iron‐dependent form of cell death, which is implicated in various diseases including cancers. However, the influence of ferroptosis‐related genes on the prognosis of breast cancer remains unclear. Methods RNA sequencing data of 1053 breast cancer tissue samples and 111 normal tissue samples from The Cancer Genome Atlas (TCGA) were analyzed. Expression levels of 259 ferroptosis‐related genes were compared. Gene Ontology (GO) and the Kyoto Gene and Genomic Encyclopedia (KEGG) analyses were conducted on differentially expressed genes. Cox univariate analysis was conducted to explore the potential prognostic biomarkers of breast cancer. Infiltrating immune cell status was assessed. Results A total of 66 ferroptosis‐related genes were differentially expressed in breast cancer tissues. The enriched GO terms included Biological Process (mainly included response to oxidative stress, cellular response to chemical stress, multicellular organismal homeostasis, cofactor metabolic process, response to metal ion, response to steroid hormone, cellular response to oxidative stress, transition metal ion homeostasis, iron ion homeostasis, and cellular iron ion homeostasis), Cellular Component (mainly included apical plasma membrane, early endosome, apical part of cell, lipid droplet, basolateral plasma membrane, blood microparticle, clathrin‐coated pit, caveola, astrocyte projection, and pronucleus) and Molecular Function (mainly included iron ion binding, ubiquitin protein ligase binding, oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, oxidoreductase activity, acting on the CH−OH group of donors, NAD or NADP as acceptor, ferric iron binding, aldo−keto reductase (NADP) activity, oxidoreductase activity, acting on single donors with incorporation of molecular oxygen, steroid dehydrogenase activity, alditol:NADP+1−oxidoreductase activity, and alcohol dehydrogenase (NADP+) activity). The enriched KEGG pathway mainly included the HIF‐1 signaling pathway, NOD‐like receptor signaling pathway, ferroptosis, IL‐17 signaling pathway, central carbon metabolism in cancer, PPAR signaling pathway, PD‐L1 expression, and PD‐1 checkpoint pathway in cancer. Among them, 38 ferroptosis‐related genes were significantly associated with the prognosis of breast cancer. The prognostic model was constructed, and breast cancer patients in low‐risk group had a better prognosis. In addition, risk score of ferroptosis prognostic model was negatively correlated with B cells (r = −0.063, p = 0.049), CD8+ T cells (r = −0.083, p = 0.010), CD4+ T cells (r = −0.097, p = 0.002), neutrophils (r = −0.068, p = 0.033), and dendritic cells (r = 0.088, p = 0.006). Conclusions The ferroptosis pathway plays a key role in breast cancer. Some differentially expressed ferroptosis‐related genes can be used as prognostic biomarkers for breast cancer.
Collapse
Affiliation(s)
- Li-Yan Jin
- Department of Thyroid and breast surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan-Lin Gu
- Department of Thyroid and breast surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhu
- Department of Thyroid and breast surgery, Traditional Chinese medicine hospital of kunshan, Kunshan, China
| | - Xiao-Hua Li
- Department of Thyroid and breast surgery, Wuzhong People's Hospital of Suzhou City, Suzhou, China
| | - Guo-Qin Jiang
- Department of Thyroid and breast surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
39
|
Redox Imbalance and Methylation Disturbances in Early Childhood Obesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2207125. [PMID: 34457110 PMCID: PMC8387800 DOI: 10.1155/2021/2207125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/13/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022]
Abstract
Obesity is increasing worldwide in prepubertal children, reducing the age of onset of associated comorbidities, including type 2 diabetes. Sulfur-containing amino acids, methionine, cysteine, and their derivatives play important roles in the transmethylation and transsulfuration pathways. Dysregulation of these pathways leads to alterations in the cellular methylation patterns and an imbalanced redox state. Therefore, we tested the hypothesis that one-carbon metabolism is already dysregulated in prepubertal children with obesity. Peripheral blood was collected from 64 children, and the plasma metabolites from transmethylation and transsulfuration pathways were quantified by HPLC. The cohort was stratified by BMI z-scores and HOMA-IR indices into healthy lean (HL), healthy obese (HO), and unhealthy obese (UHO). Fasting insulin levels were higher in the HO group compared to the HL, while the UHO had the highest. All groups presented normal fasting glycemia. Furthermore, high-density lipoprotein (HDL) was lower while triglycerides and lactate levels were higher in the UHO compared to HO subjects. S-adenosylhomocysteine (SAH) and total homocysteine levels were increased in the HO group compared to HL. Additionally, glutathione metabolism was also altered. Free cystine and oxidized glutathione (GSSG) were increased in the HO as compared to HL subjects. Importantly, the adipocyte secretory function was already compromised at this young age. Elevated circulating leptin and decreased adiponectin levels were observed in the UHO as compared to the HO subjects. Some of these alterations were concomitant with alterations in the DNA methylation patterns in the obese group, independent of the impaired insulin levels. In conclusion, our study informs on novel and important metabolic alterations in the transmethylation and the transsulfuration pathways in the early stages of obesity. Moreover, the altered secretory function of the adipocyte very early in life may be relevant in identifying early metabolic markers of disease that may inform on the increased risk for specific future comorbidities in this population.
Collapse
|
40
|
Metallocenyl derivatives of ebselen are selective and competitive inhibitors of thioredoxin reductase. J Organomet Chem 2021. [DOI: 10.1016/j.jorganchem.2021.121822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
41
|
Zuo L, Wijegunawardana D. Redox Role of ROS and Inflammation in Pulmonary Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:187-204. [PMID: 34019270 DOI: 10.1007/978-3-030-68748-9_11] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reactive oxygen species (ROS), either derived from exogenous sources or overproduced endogenously, can disrupt the body's antioxidant defenses leading to compromised redox homeostasis. The lungs are highly susceptible to ROS-mediated damage. Oxidative stress (OS) caused by this redox imbalance leads to the pathogenesis of multiple pulmonary diseases such as asthma, chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome (ARDS). OS causes damage to important cellular components in terms of lipid peroxidation, protein oxidation, and DNA histone modification. Inflammation further enhances ROS production inducing changes in transcriptional factors which mediate cellular stress response pathways. This deviation from normal cell function contributes to the detrimental pathological characteristics often seen in pulmonary diseases. Although antioxidant therapies are feasible approaches in alleviating OS-related lung impairment, a comprehensive understanding of the updated role of ROS in pulmonary inflammation is vital for the development of optimal treatments. In this chapter, we review the major pulmonary diseases-including COPD, asthma, ARDS, COVID-19, and lung cancer-as well as their association with ROS.
Collapse
Affiliation(s)
- Li Zuo
- College of Arts and Sciences, Molecular Physiology and Biophysics Lab, University of Maine, Presque Isle Campus, Presque Isle, ME, USA. .,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA.
| | | |
Collapse
|
42
|
Manuck TA, Eaves LA, Rager JE, Fry RC. Mid-pregnancy maternal blood nitric oxide-related gene and miRNA expression are associated with preterm birth. Epigenomics 2021; 13:667-682. [PMID: 33890487 DOI: 10.2217/epi-2020-0346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The nitric oxide (NO) pathway modulates inflammation and may influence birth timing. Patients & methods: Case-control analysis of 136 pregnant women with RNA obtained <28 weeks; n = 212 mRNAs and n = 108 miRNAs in the NO pathway were evaluated. NO-pathway mRNA and miRNA transcript counts in women delivering preterm versus at term were compared, miRNA-mRNA expression levels correlated and prediction models generated. Results: Fourteen genes were differentially expressed in women delivering <37 weeks; 13/14 were also differentially expressed in those delivering <34 weeks (q <0.10) versus term births. Multiple miRNA-mRNA pairs were correlated. Models with gene expression better predicted prematurity than models with only clinical or nongenomic predictors. Conclusion: Maternal blood NO pathway-related mRNA and miRNA expression is associated with prematurity.
Collapse
Affiliation(s)
- Tracy A Manuck
- Department of Obstetrics & Gynecology, Division of Maternal Fetal Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lauren A Eaves
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Julia E Rager
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rebecca C Fry
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
43
|
Identification of Redox-Sensitive Transcription Factors as Markers of Malignant Pleural Mesothelioma. Cancers (Basel) 2021; 13:cancers13051138. [PMID: 33799965 PMCID: PMC7961847 DOI: 10.3390/cancers13051138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Malignant pleural mesothelioma is a lung tumor associated with asbestos exposure, with a poor prognosis, and a difficult pharmacological approach. Asbestos exposure is very toxic for the lungs, which counteract this toxic effect by activating some antioxidant defense proteins. When these proteins are more active that in normal conditions, as in several cancers, these tumors become able to survive and resist to stress or chemotherapy. In our laboratory, we collected cellular samples of mesothelioma and non-transformed mesothelium from Hospital’s Biobank and we evaluated these proteins. Our results demonstrated these proteins are upregulated in mesothelioma cells and not in non-transformed mesothelium. This event could be associated to toxic effects evoked by asbestos exposure, highlighting the need in the future to monitor asbestos-exposed people by measuring biomarkers identified, in the attempt to identify them as possible predictive markers and potential pharmacological targets addressed to improve mesothelioma prognosis. Abstract Although asbestos has been banned in most countries around the world, malignant pleural mesothelioma (MPM) is a current problem. MPM is an aggressive tumor with a poor prognosis, so it is crucial to identify new markers in the preventive field. Asbestos exposure induces oxidative stress and its carcinogenesis has been linked to a strong oxidative damage, event counteracted by antioxidant systems at the pulmonary level. The present study has been focused on some redox-sensitive transcription factors that regulate cellular antioxidant defense and are overexpressed in many tumors, such as Nrf2 (Nuclear factor erythroid 2-related factor 2), Ref-1 (Redox effector factor 1), and FOXM1 (Forkhead box protein M1). The research was performed in human mesothelial and MPM cells. Our results have clearly demonstrated an overexpression of Nrf2, Ref-1, and FOXM1 in mesothelioma towards mesothelium, and a consequent activation of downstream genes controlled by these factors, which in turn regulates antioxidant defense. This event is mediated by oxidative free radicals produced when mesothelial cells are exposed to asbestos fibers. We observed an increased expression of Nrf2, Ref-1, and FOXM1 towards untreated cells, confirming asbestos as the mediator of oxidative stress evoked at the mesothelium level. These factors can therefore be considered predictive biomarkers of MPM and potential pharmacological targets in the treatment of this aggressive cancer.
Collapse
|
44
|
Yang Y, Neo SY, Chen Z, Cui W, Chen Y, Guo M, Wang Y, Xu H, Kurzay A, Alici E, Holmgren L, Haglund F, Wang K, Lundqvist A. Thioredoxin activity confers resistance against oxidative stress in tumor-infiltrating NK cells. J Clin Invest 2021; 130:5508-5522. [PMID: 32673292 DOI: 10.1172/jci137585] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
To improve the clinical outcome of adoptive NK cell therapy in patients with solid tumors, NK cells need to persist within the tumor microenvironment (TME) in which the abundance of ROS could dampen antitumor immune responses. In the present study, we demonstrated that IL-15-primed NK cells acquired resistance against oxidative stress through the thioredoxin system activated by mTOR. Mechanistically, the activation of thioredoxin showed dependence on localization of thioredoxin-interacting protein. We show that NK cells residing in the tumor core expressed higher thiol densities that could aid in protecting other lymphocytes against ROS within the TME. Furthermore, the prognostic value of IL15 and the NK cell gene signature in tumors may be influenced by tobacco smoking history in patients with non-small-cell lung cancer (NSCLC). Collectively, the levels of reducing antioxidants in NK cells may not only predict better tumor penetrance but potentially even the immune therapy response.
Collapse
Affiliation(s)
- Ying Yang
- School of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Shi Yong Neo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Weiyingqi Cui
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yi Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Min Guo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yongfang Wang
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyan Xu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Annina Kurzay
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Evren Alici
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Lars Holmgren
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Felix Haglund
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kai Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
In Vitro Evaluation of Antioxidant Potential of the Invasive Seagrass Halophila stipulacea. Mar Drugs 2021; 19:md19010037. [PMID: 33467094 PMCID: PMC7830009 DOI: 10.3390/md19010037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Marine organisms with fast growth rates and great biological adaptive capacity might have biotechnological interests, since ecological competitiveness might rely on enhanced physiological or biochemical processes’ capability promoting protection, defense, or repair intracellular damages. The invasive seagrass Halophila stipulacea, a non-indigenous species widespread in the Mediterranean Sea, belongs to this category. This is the premise to investigate the biotechnological interest of this species. In this study, we investigated the antioxidant activity in vitro, both in scavenging reactive oxygen species and in repairing damages from oxidative stress on the fibroblast human cell line WI-38. Together with the biochemical analysis, the antioxidant activity was characterized by the study of the expression of oxidative stress gene in WI-38 cells in presence or absence of the H. stipulacea extract. Concomitantly, the pigment pool of the extracts, as well as their macromolecular composition was characterized. This study was done separately on mature and young leaves. Results indicated that mature leaves exerted a great activity in scavenging reactive oxygen species and repairing damages from oxidative stress in the WI-38 cell line. This activity was paralleled to an enhanced carotenoids content in the mature leaf extracts and a higher carbohydrate contribution to organic matter. Our results suggest a potential of the old leaves of H. stipulacea as oxidative stress damage protecting or repair agents in fibroblast cell lines. This study paves the way to transmute the invasive H. stipulacea environmental threat in goods for human health.
Collapse
|
46
|
Koh WX, Gomez AP, Lee J, Mohameed JBH, Leong WK. Relative reactivity of the dinuclear ruthenium complex [CpRu(CO)2]2 with diphenylselenyl sulphide and diphenyl disulphide. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
47
|
Al Zoubi M, Aljabali A. Polymorphisms, antioxidant genes, and cancer. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00010-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
48
|
Peraldo-Neia C, Ostano P, Mello-Grand M, Guana F, Gregnanin I, Boschi D, Oliaro-Bosso S, Pippione AC, Carenzo A, De Cecco L, Cavalieri S, Micali A, Perrone F, Averono G, Bagnasacco P, Dosdegani R, Masini L, Krengli M, Aluffi-Valletti P, Valente G, Chiorino G. AKR1C3 is a biomarker and druggable target for oropharyngeal tumors. Cell Oncol (Dordr) 2020; 44:357-372. [PMID: 33211282 DOI: 10.1007/s13402-020-00571-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 10/22/2022] Open
Abstract
PURPOSE Oropharynx squamous cell carcinoma (OPSCC) is a subtype of head and neck squamous cell carcinoma (HNSCC) arising from the base of the tongue, lingual tonsils, tonsils, oropharynx or pharynx. The majority of HPV-positive OPSCCs has a good prognosis, but a fraction of them has a poor prognosis, similar to HPV-negative OPSCCs. An in-depth understanding of the molecular mechanisms underlying OPSCC is mandatory for the identification of novel prognostic biomarkers and/or novel therapeutic targets. METHODS 14 HPV-positive and 15 HPV-negative OPSCCs with 5-year follow-up information were subjected to gene expression profiling and, subsequently, compared to three extensive published OPSCC cohorts to define robust biomarkers for HPV-negative lesions. Validation of Aldo-keto-reductases 1C3 (AKR1C3) by qRT-PCR was carried out on an independent cohort (n = 111) of OPSCC cases. In addition, OPSCC cell lines Fadu and Cal-27 were treated with Cisplatin and/or specific AKR1C3 inhibitors to assess their (combined) therapeutic effects. RESULTS Gene set enrichment analysis (GSEA) on the four datasets revealed that the genes down-regulated in HPV-negative samples were mainly involved in immune system, whereas those up-regulated mainly in glutathione derivative biosynthetic and xenobiotic metabolic processes. A panel of 30 robust HPV-associated transcripts was identified, with AKR1C3 as top-overexpressed transcript in HPV-negative samples. AKR1C3 expression in 111 independent OPSCC cases positively correlated with a worse survival, both in the entire cohort and in HPV-positive samples. Pretreatment with a selective AKR1C3 inhibitor potentiated the effect of Cisplatin in OPSCC cells exhibiting higher basal AKR1C3 expression levels. CONCLUSIONS We identified AKR1C3 as a potential prognostic biomarker in OPSCC and as a potential drug target whose inhibition can potentiate the effect of Cisplatin.
Collapse
Affiliation(s)
- Caterina Peraldo-Neia
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Paola Ostano
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Maurizia Mello-Grand
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Francesca Guana
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Ilaria Gregnanin
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Donatella Boschi
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Simonetta Oliaro-Bosso
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Agnese Chiara Pippione
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Andrea Carenzo
- Integrated Biology Platform, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Loris De Cecco
- Integrated Biology Platform, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Arianna Micali
- Integrated Biology Platform, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Federica Perrone
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Gianluca Averono
- Otorhinolaryngology Unit, Ospedale degli Infermi, via dei Ponderanesi 1, Ponderano, Biella, Italy
| | - Paolo Bagnasacco
- Otorhinolaryngology Unit, Ospedale degli Infermi, via dei Ponderanesi 1, Ponderano, Biella, Italy
| | | | - Laura Masini
- Department of Translational Medicine, UPO School of Medicine, Radiotherapy Unit, Novara, Italy
| | - Marco Krengli
- Department of Translational Medicine, UPO School of Medicine, Radiotherapy Unit, Novara, Italy
| | - Paolo Aluffi-Valletti
- Department of Health Sciences, UPO School of Medicine, Otorhinolaryngology Unit, Novara, Italy
| | - Guido Valente
- Department of Translational Medicine, UPO School of Medicine, Radiotherapy Unit, Novara, Italy
| | - Giovanna Chiorino
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy.
| |
Collapse
|
49
|
Lin W, Lai Y, Kalyanam N, Ho C, Pan M. S
‐Allylcysteine Inhibits PhIP/DSS‐Induced Colon Carcinogenesis through Mitigating Inflammation, Targeting Keap1, and Modulating Microbiota Composition in Mice. Mol Nutr Food Res 2020. [DOI: 10.1002/mnfr.202000576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Wei‐Sheng Lin
- Institute of Food Science and Technology National Taiwan University Taipei 10617 Taiwan
| | - Ying‐Jang Lai
- Department of Food Science National Quemoy University Quemoy County 89250 Taiwan
| | | | - Chi‐Tang Ho
- Department of Food Science Rutgers University New Brunswick NJ 08901 USA
| | - Min‐Hsiung Pan
- Institute of Food Science and Technology National Taiwan University Taipei 10617 Taiwan
- Department of Medical Research China Medical University Hospital China Medical University Taichung 40402 Taiwan
- Department of Health and Nutrition Biotechnology Asia University Taichung 41354 Taiwan
| |
Collapse
|
50
|
Jovanović M, Dragoj M, Zhukovsky D, Dar'in D, Krasavin M, Pešić M, Podolski-Renić A. Novel TrxR1 Inhibitors Show Potential for Glioma Treatment by Suppressing the Invasion and Sensitizing Glioma Cells to Chemotherapy. Front Mol Biosci 2020; 7:586146. [PMID: 33134322 PMCID: PMC7573255 DOI: 10.3389/fmolb.2020.586146] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/10/2020] [Indexed: 01/13/2023] Open
Abstract
Currently, available glioblastoma (GBM) treatment remains ineffective, with relapse after initial response and low survival rate of GBM patients. The reasons behind limited capacities for GBM treatment are high tumor heterogeneity, invasiveness, and occurrence of drug resistance. Therefore, developing novel therapeutic strategies is of utmost importance. Thioredoxin reductase (TrxR) is a novel, promising target due to its overexpression in many cancer types and important role in cancer progression. Previous research on Ugi-type Michael acceptors–inhibitors of TrxR showed desirable anticancer properties, with significant selectivity toward cancer cells. Herein, two TrxR inhibitors, 5 and 6, underwent in-depth study on multidrug-resistant (MDR) glioma cell lines. Besides the antioxidative effects, 5 and 6 induced cell death, decreased cell proliferation, and suppressed invasion and migration of glioma cells. Both compounds showed a synergistic effect in combination with temozolomide (TMZ), a first-line chemotherapeutic for GBM treatment. Moreover, 5 and 6 affected activity of P-glycoprotein extrusion pump that could be found in cancer cells and in the blood–brain barrier (BBB), thus showing potential for suppressing MDR phenotype in cancer cells and evading BBB. In conclusion, investigated TrxR inhibitors are effective anticancer compounds, acting through inhibition of the thioredoxin system and perturbation of antioxidative defense systems of glioma cells. They are suitable for combining with other chemotherapeutics, able to surpass the BBB and overcome MDR. Thus, our findings suggest further exploration of Ugi-type Michael acceptors–TrxR inhibitors’ potential as an adjuvant therapy for GBM treatment.
Collapse
Affiliation(s)
- Mirna Jovanović
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miodrag Dragoj
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Daniil Zhukovsky
- Institute of Chemistry, Saint Petersburg State University, Russian Federation, Saint Petersburg, Russia
| | - Dmitry Dar'in
- Institute of Chemistry, Saint Petersburg State University, Russian Federation, Saint Petersburg, Russia
| | - Mikhail Krasavin
- Institute of Chemistry, Saint Petersburg State University, Russian Federation, Saint Petersburg, Russia
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|